Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EBioMedicine ; 30: 129-137, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29574092

RESUMO

BACKGROUND: Biomarker-targeted molecular imaging holds promise for early detection of pancreatic cancer. The aim of this study was to design and evaluate a plectin-1 targeted multi-functional nanoparticle probe for pancreatic cancer imaging. METHODS: 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-amino(polyethylene glycol) (DSPE-PEG-NH2)-modified superparamagnetic iron oxide (Fe3O4) nanoparticles (SPION) were conjugated with plectin-1 antibody and/or Cy7 to create the multi-functional targeted nanoparticle targeted probe (Plectin-SPION-Cy7) or non-targeted probe (SPION-Cy7). Pancreatic carcinoma cell lines expressing plectin-1 were cultured with the targeted or control probes and then were imaged using confocal laser scanning microscopy and magnetic resonance imaging (MRI). Accumulations of the nanoparticles in pancreatic tumor xenografted mice were determined by MRI and fluorescence imaging. RESULTS: In vitro optical imaging and MRI showed that the targeted nanoparticles were highly accumulated in MIAPaCa2 and XPA-1 carcinoma cells but not in non-carcinoma MIN6 cells, which was further confirmed by Prussian blue staining. In vivo MRI showed a significant T2 signal reduction. Prussian blue staining further confirmed that the plectin-1 targeted nanoparticles were highly accumulated in the tumor mass but not in normal pancreatic tissues, or in the liver and kidney, and few nanoparticles were observed in the tumors of mice injected with SPION-Cy7. CONCLUSIONS: Our data demonstrate that plectin-1 targeted fluorescence and MR dual-functional nanoparticle can visualize pancreatic cancer, and it has great potential to be used with various imaging devices for pancreatic cancer detection.


Assuntos
Imageamento por Ressonância Magnética , Nanopartículas/química , Neoplasias Pancreáticas/diagnóstico , Plectina/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Magnetismo , Camundongos Nus , Nanopartículas/ultraestrutura , Imagem Óptica , Neoplasias Pancreáticas/patologia
2.
Chin J Integr Med ; 2016 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-27358203

RESUMO

OBJECTIVE: To further explore the anti-cancer effect of Tounong Powder () extracts (TNSEs) on human colon cancer LoVo cells and examine the possible molecular mechanisms. METHODS: The contents of TNSEs were determined by liquid chromatograph-mass spectrometer (LC-MS) analysis after extraction with water and methanol. Variations of cell morphological features were observed using fluorescence microscopy. Cytotoxicity was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell cycle distribution and apoptosis were analyzed using flow cytometry at different TNSE doses (0, 62.5, 125, or 250 µg/mL). Protein expressions of phosphatidylinositol 3-kinase (PI3K), phosphate protein kinase B (p-AKT), phosphate mammalian target of rapamycin (p-mTOR), p-p70s6k1, cleaved caspase-9 and -3 were detected using Western blot analysis. RESULTS: TNSEs induced cell growth inhibition in a concentration- and time-dependent manner. Flow cytometric analysis showed apoptotic cells and cell cycle arrest at the G phase after TNSEs treatment compared with controls. Furthermore, TNSEs significantly down-regulated the proteins PI3K, p-AKT, p-mTOR, and p-p70s6k1, and up-regulated the proteins cleaved caspase-9 and -3 dosedependently, as determined by Western blot. CONCLUSIONS: TNSEs reduced LoVo cell proliferation, and caused apoptosis and cell-cycle arrest in LoVo cells. This effect might be associated with regulation of the PI3K/AKT signaling pathway.

3.
Mol Clin Oncol ; 4(6): 929-932, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27284425

RESUMO

Leukocytoclastic vasculitis (LCV) is a neutrophilic inflammation of the blood vessels. LCV may present as a paraneoplastic syndrome occurring before, synchronously with, or after the diagnosis of malignancy. In this study, we report a unique case of multiple malignancies developing simultaneously in a patient with a long history of LCV. The patient was originally diagnosed with LCV and received long-term glucocorticoid treatment. After 11 years of therapy, the patient developed three primary malignancies, including small-cell lung carcinoma, gastric adenocarcinoma and colonic adenocarcinoma. It is likely that LCV was not a paraneoplastic syndrome in this case, but rather an independent process, and the development of multiple cancers is likely associated with the long-term glucocorticoid treatment, which caused imbalance of the immune system. Although the development of cancer during the course of glucocorticoid treatment is very rare, clinicians must be aware of this possible association and immunodysregulation may play a role in this context.

4.
Nanomedicine ; 12(6): 1651-61, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26995092

RESUMO

We investigated the potential of targeting survivin, an inhibitor of apoptosis, in visualize pancreatic tumor in mouse model using targeted magnetic nanoparticles (MNPs) and magnetic resonance imaging (MRI). Chitosan-coated MNPS and survivin antisense oligonucleotide(ASON) were conjugated to give Sur-MNPs. Accumulations of targeted, non-targeted nanoparticles or nonsense oligonucleotide-MNPs (NSON-MNPs) in the liver, spleen, kidney and tumors were determined. Targeted nanoparticles were highly accumulated in BxPC-3 cells but not in non-cancer cells. In vivo MRI showed a significant T2 signal reduction in tumors of mice injected with targeted nanoparticles but slight signal change in tumors of mice injected with non-targeted nanoparticles or NSON-MNPs. Prussian blue staining demonstrated highly accumulated Sur-MNPs in tumor mass compared with normal pancreatic, kidney and liver tissues. Our data show that the MNPs functionalized with ASON lead to the targeted localization in pancreatic tumors. Survivin targeted nanoparticles could be used for detection of pancreatic tumors.


Assuntos
Proteínas Inibidoras de Apoptose/efeitos dos fármacos , Nanopartículas de Magnetita/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Repressoras/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Imageamento por Ressonância Magnética , Camundongos , Survivina
5.
PLoS One ; 10(7): e0132799, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26181041

RESUMO

Relapsed, refractory lymphoma remains to be a challenge and lacks efficient treatment. Some tumor cells escape from treatment, become resistant to chemotherapeutic agents, and rapidly regenerate into large tumors. Lymphoma cells induce accumulation of Gr-1(+-)CD11b(+) myeloid derived suppressor cells (MDSCs) in lymphatic organs and their vicinity. MDSCs enable tumor cells to escape from immune cells mediated surveillance and attack. Gemcitabine is a chemotherapeutic agent that eliminates both tumor cells and MDSCs, improving the immune environment favorable for subsequent treatment. We evaluated the effects of low dose gemcitabine combined with intra-tumorally delivered dendritic cells (DCs) for the treatment of A20 large-size lymphoma. We showed that MDSCs increased markedly in lymphoma-bearing mice, and that gemcitabine significantly increased the apoptosis of MDSCs. Treatment of lymphoma with either gemcitabine or intra-tumoral DCs alone could not inhibit tumor growth or rescue lymphoma-bearing mice. Treatment of lymphoma with small dose gemcitabine followed by intra-tumorally injected DCs significantly improved the efficacy of either individual treatment by reducing MDSCs, inducing onsite DCs maturation, eliminating tumor cells, inhibiting tumor growth and relapse, and extending the survival of the lymphoma-bearing mice, partly through the induction of the IFNγ secreting cells and the activation of cytotoxic lymphocytes. We showed that NK cells and CD8(+ )T cells were the major effectors to mediate the inhibition of tumor growth. Thus, the observation that gemcitabine synergizes DCs mediated immunotherapy to improve the efficacy of large size lymphoma treatment provides an experimental basis for the combination of chemotherapy and immunotherapy for the efficient treatment of relapsed or refractory lymphoma.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Células Dendríticas/transplante , Desoxicitidina/análogos & derivados , Linfoma de Células B/terapia , Células Mieloides/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Terapia Combinada , Citotoxicidade Imunológica , Células Dendríticas/citologia , Células Dendríticas/imunologia , Desoxicitidina/farmacologia , Progressão da Doença , Humanos , Imunoterapia/métodos , Injeções Intralesionais , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Linfoma de Células B/imunologia , Linfoma de Células B/mortalidade , Linfoma de Células B/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/imunologia , Células Mieloides/patologia , Análise de Sobrevida , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
6.
Artigo em Inglês | MEDLINE | ID: mdl-25788964

RESUMO

Tou Nong San (TNS) is a traditional Chinese medicinal decoction used to treat sores and carbuncles. It contains four herbal drugs and one animal medicine: Radix Astragaliseu Seu Hedysari, Angelica sinensis, Ligustici Chuanxiong, Spina Gleditsiae, and stir-baked Squama Manis. Previous studies have shown that it has anticancer effects. This report validates in vivo antitumor properties of TNS. The compounds contained in TNSE were confirmed by liquid chromatographmass spectrometer (LC-MS) analysis. The in vivo antitumor activity of TNS extract (TNSE) was tested by feeding it to athymic mice harboring a human colonic tumor subcutaneous xenograft. Toxicity was monitored by recording behavior and weight parameters. Seven compounds were detected in TNSE by LC-MS. TNSE was fed to athymic mice for 2 weeks. No adverse reactions were reported. Compared to the control group, administration of TNSE to tumor bearing mice significantly reduced both tumor weight and volume. The expressions of p-PI3K, p-AKT, p-mTOR, p-p70s6k1, VEGF, and CD31 were significantly reduced, the expression levels of cleaved Caspase-9 and cleaved Caspase-3 were significantly increased in the TNSE groups compared to the control group as determined by western blot and immunohistochemistry. TNSE produced anticolonic cancer effects and the underlying mechanisms involved inhibition of the PI3K/AKT signal transduction pathway, inhibition of angiogenesis, and promotion of apoptotic proteins.

7.
Biomarkers ; 19(1): 9-15, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24237184

RESUMO

Transforming growth factor-alpha (TGF-α) correlates with deep invasion, advanced stage and poor prognosis in gastric cancer. Genetic variants in the 3' untranslated region (UTR) of TGF-α gene may influence the stability and post-transcriptional regulation of mRNA and contribute to gastric cancer predisposition. To test this hypothesis, we genotyped five polymorphisms in 3'UTR (rs3771527, rs503314, rs473698, rs3732253 and rs538118) and one in 3' near region (rs11466306) of the TGF-α gene by polymerase chain reaction-ligation detection reaction methods (PCR-LDR). We found that GA/AA genotype of rs11466306 in the 3' near gene could increase the risk of overall gastric cancer (adjusted OR = 1.499, 95%CI: 1.101-2.041), compared to the wild homozygous GG genotype. Meanwhile, the risk effect was more obvious in the intestinal gastric cancer and gastric noncardia cancer (adjusted OR = 1.682, 95%CI: 1.188-2.380; adjusted OR = 1.495, 95%CI: 1.072-2.086, respectively), but not for the diffuse type and gastric cardia cancer (p > 0.05). CT/TT genotype for rs3732253 in the 3' UTR was associated with increased risk of intestinal gastric cancer (adjusted OR = 1.464, 95%CI: 1.036-2.069), compared to their wild homozygous genotypes. These findings indicate that potentially functional TGF-α gene variant may contribute to the risk of intestinal gastric cancer and/or gastric noncardia cancer and could be used as molecular markers for detecting intestinal gastric cancer and/or gastric noncardia cancer in Chinese Han population.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Gástricas/genética , Fator de Crescimento Transformador alfa/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , China , Feminino , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Risco , Neoplasias Gástricas/patologia
8.
Int J Nanomedicine ; 7: 4109-18, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22888247

RESUMO

BACKGROUND: The present study evaluated whether magnetic nanoparticles containing Fe(3)O(4) could enhance the activity of gambogic acid in human colon cancer cells, and explored the potential mechanisms involved. METHODS: Cytotoxicity was evaluated by MTT assay. The percentage of cells undergoing apoptosis was analyzed by flow cytometry, and cell morphology was observed under both an optical microscope and a fluorescence microscope. Reverse transcriptase polymerase chain reaction and Western blot assay were performed to determine the transcription of genes and expression of proteins, respectively. RESULTS: Gambogic acid could inhibit proliferation of LOVO cells in a dose-dependent and time-dependent manner and induce apoptosis, which was dramatically enhanced by magnetic nanoparticles containing Fe(3)O(4). The typical morphological features of apoptosis in LOVO cells were observed after treatment comprising gambogic acid with and without magnetic nanoparticles containing Fe(3)O(4). Transcription of cytochrome c, caspase 9, and caspase 3 genes was higher in the group treated with magnetic nanoparticles containing Fe(3)O(4) and gambogic acid than in the groups that received gambogic acid or magnetic nanoparticles containing Fe(3)O(4), but transcription of phosphatidylinositol 3-kinase, Akt, and Bad genes decreased. Notably, expression of cytochrome c, caspase 9, and caspase 3 proteins in the group treated with gambogic acid and magnetic nanoparticles containing Fe(3)O(4) was higher than in the groups receiving magnetic nanoparticles containing Fe(3)O(4) or gambogic acid, while expression of p-PI3K, p-Akt, p-Bad, pro-caspase 9, and pro-caspase 3 degraded. CONCLUSION: Magnetic nanoparticles containing Fe(3)O(4) can enhance apoptosis induced by gambogic acid which may be closely related to regulation of the PI3K/Akt/Bad pathway in the treatment of human colon cancer.


Assuntos
Nanopartículas de Magnetita/administração & dosagem , Nanopartículas de Magnetita/química , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Xantonas/farmacologia , Proteína de Morte Celular Associada a bcl/metabolismo , Análise de Variância , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Citometria de Fluxo , Humanos , Fosfatidilinositol 3-Quinase/genética , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/efeitos dos fármacos , Xantonas/química , Proteína de Morte Celular Associada a bcl/genética
9.
Chin J Integr Med ; 18(7): 522-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22772915

RESUMO

OBJECTIVE: To investigate the effects of Tounongsan () extract (TNSE) on proliferation and apoptosis of the human lymphoma cell line Raji and its possible mechanism of action. METHODS: The viability of TNSE-treated Raji cells was measured by a 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Cell apoptosis was determined by flow cytometry. The molecular mechanisms of TNSE-mediated apoptosis were further investigated by reverse transcription-polymerase chain reaction (RT-PCR) analysis of the mRNA expression of nuclear factor κB (NF-κB), Bcl-xL, Bcl-2-associated death promoter (Bad), caspase-9 and caspase-3. Western blotting was used to detect the protein expressions of NF-κB, Bad, cleaved caspase-9 and cleaved caspase-3. RESULTS: TNSE inhibited Raji cell proliferation in dose- and time-dependent manners. After 48-h treatment with various concentrations of TNSE (125, 250 and 500 µg/mL), the apoptosis rates of Raji cell were 12.23%±1.98% (P<0.05), 20.97%±3.96% (P<0.01) and 30.4%±4.87% (P<0.01), respectively, compared with those of the control (6.02%±1.01%). RT-PCR demonstrated that NF-κB mRNA expression was significantly downregulated in Raji cells treated with 250 µg/mL TNSE for 48 h (P<0.05), while Bad, caspase-9 and caspase-3 mRNA levels were upregulated (P<0.05). Moreover, TNSE treatment resulted in downregulation of NF-κB protein expression and strikingly upregulated protein expressions of Bad, cleaved caspase-9, cleaved caspase-3 in a dose-dependent manner, as determined by Western blot. CONCLUSION: TNSE exhibits significant anti-proliferative and apoptotic effects in Raji cells, which may be involved in regulation of NF-κB and Bad, and activation of caspase-9 and caspase-3.


Assuntos
Apoptose/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Extratos Vegetais/farmacologia , Caspase 3/genética , Caspase 3/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , NF-kappa B/genética , NF-kappa B/metabolismo , Células Tumorais Cultivadas , Proteína de Morte Celular Associada a bcl/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...